Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Nat Immunol ; 10(4): 348-55, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19295631

RESUMO

Death receptors (DRs) are members of the tumor necrosis factor receptor superfamily that possess a cytoplasmic death domain (DD). DRs regulate important operational and homeostatic aspects of the immune system. They transmit signals through apical protein complexes, which are nucleated by the DD adaptors FADD and TRADD, to control cellular outcomes that range from apoptosis to gene activation. FADD and TRADD also nucleate several distal signaling complexes, which mediate cross-talk between distinct DR signaling pathways. Moreover, together with other DR signal transducers, FADD and TRADD participate in functional complexes assembled by certain non-DR immune cell receptors, such as pattern-recognition receptors. Thus, DR signal transducers may provide important nodes of coordination in immune signaling networks.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Proteína de Domínio de Morte Associada a Fas/fisiologia , Receptores de Morte Celular/fisiologia , Transdução de Sinais/fisiologia , Proteína de Domínio de Morte Associada a Receptor de TNF/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/imunologia , Animais , Apoptose/fisiologia , Proteína de Domínio de Morte Associada a Fas/imunologia , Humanos , Imunidade Ativa , Imunidade Inata , Receptores de Morte Celular/imunologia , Receptores do Fator de Necrose Tumoral/imunologia , Receptores do Fator de Necrose Tumoral/fisiologia , Proteína de Domínio de Morte Associada a Receptor de TNF/imunologia
2.
Trends Immunol ; 39(12): 1036-1053, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30401514

RESUMO

Initially described as an adaptor molecule for death receptor (DR)-mediated apoptosis, Fas-associated death domain (FADD) was later implicated in nonapoptotic cellular processes. During the last decade, FADD has been shown to participate and regulate most of the signalosome complexes, including necrosome, FADDosome, innateosome, and inflammasome. Given the role of these signaling complexes, FADD has emerged as a new actor in innate immunity, inflammation, and cancer development. Concomitant to these new roles, a surprising number of mechanisms deemed to regulate FADD functions have been identified, including post-translational modifications of FADD protein and FADD secretion. This review focuses on recent knowledge of the biological roles of FADD, a pleiotropic molecule having multiple partners, and its impact in cancer, innate immunity, and inflammation.


Assuntos
Proteína de Domínio de Morte Associada a Fas/metabolismo , Inflamação/metabolismo , Neoplasias/metabolismo , Animais , Domínio de Morte , Proteína de Domínio de Morte Associada a Fas/imunologia , Humanos , Inflamação/imunologia , Neoplasias/imunologia
3.
Immunity ; 35(4): 572-82, 2011 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-22000287

RESUMO

Epidermal keratinocytes provide an essential structural and immunological barrier forming the first line of defense against potentially pathogenic microorganisms. Mechanisms regulating barrier integrity and innate immune responses in the epidermis are important for the maintenance of skin immune homeostasis and the pathogenesis of inflammatory skin diseases. Here, we show that epidermal keratinocyte-restricted deficiency of the adaptor protein FADD (FADD(E-KO)) induced severe inflammatory skin lesions in mice. The development of skin inflammation in FADD(E-KO) mice was triggered by RIP kinase 3 (RIP3)-mediated programmed necrosis (termed necroptosis) of FADD-deficient keratinocytes, which was partly dependent on the deubiquitinating enzyme CYLD and tumor necrosis factor (TNF)-TNF receptor 1 signaling. Collectively, our findings provide an in vivo experimental paradigm that regulation of necroptosis in keratinocytes is important for the maintenance of immune homeostasis and the prevention of chronic inflammation in the skin.


Assuntos
Apoptose , Dermatite/imunologia , Dermatite/patologia , Epiderme/imunologia , Proteína de Domínio de Morte Associada a Fas/imunologia , Queratinócitos/imunologia , Animais , Células Cultivadas , Dermatite/metabolismo , Proteína de Domínio de Morte Associada a Fas/deficiência , Homeostase , Camundongos , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/metabolismo , Necrose , Transdução de Sinais
4.
Fish Shellfish Immunol ; 89: 548-554, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30991146

RESUMO

In this study, a sea cucumber Fas-associated death domain (FADD) named HLFADD was first cloned from Holothuria leucospilota. The full-length cDNA of HLFADD is 2137 bp in size, containing a 116-bp 5'-untranslated region (UTR), a 1334-bp 3'-UTR and a 687-bp open reading frame (ORF) encoding a protein of 228 amino acids with a deduced molecular weight of 26.42 kDa. HLFADD protein contains a conserved death effector domain at its N-terminal and a conserved death domain at its C-terminal, structurally similar to its counterparts in vertebrates. The over-expressed HLFADD protein could induce apoptosis in HEK293 cells, suggesting a possible death receptor-mediated apoptosis pathway in echinoderms adapted with FADD. Moreover, HLFADD mRNA is ubiquitously expressed in all examined tissues, with the highest transcript level in the coelomocytes, followed by intestine. In vitro experiments performed in the H. leucospilota coelomocytes, the expression of HLFADD mRNA was significantly up-regulated by lipopolysaccharides (LPS) or polyriboinosinic-polyribocytidylic acid [poly (I:C)] challenge, suggesting that HLFADD might play important roles in the innate immune defense of sea cucumber against the invasion of bacteria and viruses.


Assuntos
Proteína de Domínio de Morte Associada a Fas/genética , Proteína de Domínio de Morte Associada a Fas/imunologia , Regulação da Expressão Gênica/imunologia , Holothuria/genética , Holothuria/imunologia , Imunidade Inata/genética , Sequência de Aminoácidos , Animais , Apoptose , Sequência de Bases , Proteína de Domínio de Morte Associada a Fas/química , Perfilação da Expressão Gênica , Células HEK293 , Humanos , Lipopolissacarídeos/farmacologia , Filogenia , Poli I-C/farmacologia , Alinhamento de Sequência , Regulação para Cima
5.
Fish Shellfish Immunol ; 74: 517-529, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29355760

RESUMO

Fas-associated protein with death domain (FADD) is the key adaptor protein that transmits apoptotic signals mediated by the main death receptors. Besides being an essential instrument in cell death, FADD is also implicated in proliferation, cell cycle progression, tumor development, inflammation, innate immunity, and autophagy. In the present study, a FADD homologue (EcFADD) from the orange-spotted grouper (Epinephelus coioides) was cloned and its possible role in fish immunity was analyzed. The full length cDNA of EcFADD contains 808 base pairs (bp), including a 573 bp open reading frame that encodes a 190 amino acid protein with a predicted molecular mass of 21.81 kDa. Quantitative real-time polymerase chain reaction analysis indicated that EcFADD was distributed in all examined tissues. The expression of EcFADD in the spleen of E. coioides was differentially up-regulated when challenged with Singapore grouper iridovirus (SGIV) or polyinosine-polycytidylic acid(poly[I:C]). EcFADD was abundantly distributed in both the cytoplasm and nucleus in grouper spleen (GS) and fathead minnow (FHM) epithelial cells. Over-expression of EcFADD inhibited SGIV infection and replication and SGIV-induced apoptosis. To achieve antiviral and anti-apoptosis activities, FADD promoted the activation of interferon-stimulated response element (ISRE) and type I interferon (IFN) genes in the antiviral IFN signaling pathway and inhibited activation of apoptosis-related transcription factors p53. Our results not only characterize FADD but also reveal new immune functions and the molecular mechanisms by which FADD responds to virus infection and virus-induced apoptosis.


Assuntos
Bass/genética , Bass/imunologia , Proteína de Domínio de Morte Associada a Fas/genética , Proteína de Domínio de Morte Associada a Fas/imunologia , Doenças dos Peixes/imunologia , Regulação da Expressão Gênica/imunologia , Imunidade Inata/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Proteína de Domínio de Morte Associada a Fas/química , Proteínas de Peixes/química , Proteínas de Peixes/genética , Proteínas de Peixes/imunologia , Perfilação da Expressão Gênica , Filogenia , Poli I-C/farmacologia , Ranavirus/fisiologia , Alinhamento de Sequência/veterinária
6.
J Immunol ; 196(1): 407-15, 2016 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-26582950

RESUMO

The cytokine IL-1ß is intimately linked to many pathological inflammatory conditions. Mature IL-1ß secretion requires cleavage by the inflammasome. Recent evidence indicates that many cell death signal adaptors have regulatory roles in inflammasome activity. These include the apoptosis inducers FADD and caspase 8, and the necroptosis kinases receptor interacting protein kinase 1 (RIPK1) and RIPK3. PGAM5 is a mitochondrial phosphatase that has been reported to function downstream of RIPK3 to promote necroptosis and IL-1ß secretion. To interrogate the biological function of PGAM5, we generated Pgam5(-/-) mice. We found that Pgam5(-/-) mice were smaller compared with wild type littermates, and male Pgam5(-/-) mice were born at sub-Mendelian ratio. Despite these growth and survival defects, Pgam5(-/-) cells responded normally to multiple inducers of apoptosis and necroptosis. Rather, we found that PGAM5 is critical for IL-1ß secretion in response to NLRP3 and AIM2 inflammasome agonists. Moreover, vesicular stomatosis virus-induced IL-1ß secretion was impaired in Pgam5(-/-) bone marrow-derived macrophages, but not in Ripk3(-/-) bone marrow-derived dendritic cells, indicating that PGAM5 functions independent of RIPK3 to promote inflammasome activation. Mechanistically, PGAM5 promotes ASC polymerization, maintenance of mitochondrial integrity, and optimal reactive oxygen species production in response to inflammasome signals. Hence PGAM5 is a novel regulator of inflammasome and caspase 1 activity that functions independently of RIPK3.


Assuntos
Apoptose/imunologia , Inflamassomos/imunologia , Interleucina-1beta/metabolismo , Macrófagos/imunologia , Monoéster Fosfórico Hidrolases/genética , Animais , Proteínas de Transporte/imunologia , Caspase 1/imunologia , Caspase 8/imunologia , Células Cultivadas , Proteínas de Ligação a DNA/imunologia , Células Dendríticas/imunologia , Proteína de Domínio de Morte Associada a Fas/imunologia , Inflamação/imunologia , Interleucina-1beta/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/imunologia , Mitocôndrias/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR , Fosfoproteínas Fosfatases , Monoéster Fosfórico Hidrolases/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Proteína Serina-Treonina Quinases de Interação com Receptores/genética , Proteína Serina-Treonina Quinases de Interação com Receptores/imunologia , Transdução de Sinais/imunologia , Vírus da Estomatite Vesicular Indiana/imunologia
7.
Biochem Biophys Res Commun ; 485(1): 152-159, 2017 03 25.
Artigo em Inglês | MEDLINE | ID: mdl-28189684

RESUMO

NF-κB is crucial to regulate immune and inflammatory responses and cell survival. LUBAC generates a linear ubiquitin chain and activates NF-κB through ubiquitin ligase (E3) activity in the HOIP subunit. Here, we show that HOIP is predominantly cleaved by caspase at Asp390 upon apoptosis, and that is subjected to proteasomal degradation. We identified that FADD, as well as NEMO, is a substrate for LUBAC. Although the C-terminal fragment of HOIP retains NF-κB activity, linear ubiquitination of NEMO and FADD decreases upon apoptosis. Moreover, the N-terminal fragment of HOIP binds with deubiquitinases, such as OTULIN and CYLD-SPATA2. These results indicate that caspase-mediated cleavage of HOIP divides critical functional regions of HOIP, and that this regulates linear (de)ubiquitination of substrates upon apoptosis.


Assuntos
Apoptose , Caspases/metabolismo , Proteína de Domínio de Morte Associada a Fas/metabolismo , Quinase I-kappa B/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação , Caspases/imunologia , Proteína de Domínio de Morte Associada a Fas/imunologia , Técnicas de Inativação de Genes , Células HEK293 , Humanos , Quinase I-kappa B/imunologia , Células Jurkat , NF-kappa B/imunologia , NF-kappa B/metabolismo , Fator de Necrose Tumoral alfa/imunologia , Fator de Necrose Tumoral alfa/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/imunologia
8.
PLoS Pathog ; 9(10): e1003721, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24204270

RESUMO

RIG-I-Like Receptors (RLRs) sense cytosolic viral RNA to transiently activate type I IFN production. Here, we report that a type I IFN inducible DExD/H helicase, DDX24, exerts a negative-regulatory effect on RLR function. Expression of DDX24 specifically suppressed RLR activity, while DDX24 loss, which caused embryonic lethality, augmented cytosolic RNA-mediated innate signaling and facilitated RNA virus replication. DDX24 preferentially bound to RNA rather than DNA species and influenced signaling by associating with adaptor proteins FADD and RIP1. These events preferentially impeded IRF7 activity, an essential transcription factor for type I IFN production. Our data provide a new function for DDX24 and help explain innate immune gene regulation, mechanisms that may additionally provide insight into the causes of inflammatory disease.


Assuntos
RNA Helicases DEAD-box/imunologia , Imunidade Inata , RNA Viral/imunologia , Transdução de Sinais/imunologia , Estomatite Vesicular/imunologia , Vírus da Estomatite Vesicular Indiana/imunologia , Animais , RNA Helicases DEAD-box/genética , Proteína de Domínio de Morte Associada a Fas/genética , Proteína de Domínio de Morte Associada a Fas/imunologia , Proteínas Ativadoras de GTPase/genética , Proteínas Ativadoras de GTPase/imunologia , Fator Regulador 7 de Interferon/genética , Fator Regulador 7 de Interferon/imunologia , Interferon Tipo I/genética , Interferon Tipo I/imunologia , Camundongos , Camundongos Knockout , RNA Viral/genética , Transdução de Sinais/genética , Estomatite Vesicular/genética , Estomatite Vesicular/patologia , Vírus da Estomatite Vesicular Indiana/genética
9.
J Immunol ; 188(12): 6194-204, 2012 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-22611243

RESUMO

We found that an adaptor protein, signal-transducing adaptor protein (STAP)-2, is a new member of the Fas-death-inducing signaling complex and participates in activation-induced cell death in T cells. STAP-2 enhanced Fas-mediated apoptosis and caspase-8 aggregation and activation in Jurkat T cells. Importantly, STAP-2 directly interacted with caspase-8 and Fas, resulting in enhanced interactions between caspase-8 and FADD in the Fas-death-inducing signaling complex. Moreover, STAP-2 protein has a consensus caspase-8 cleavage sequence, VEAD, in its C-terminal domain, and processing of STAP-2 by caspase-8 was crucial for Fas-induced apoptosis. Physiologic roles of STAP-2 were confirmed by observations that STAP-2-deficient mice displayed impaired activation-induced cell death and superantigen-induced T cell depletion. Therefore, STAP-2 is a novel participant in the regulation of T cell apoptosis after stimulation.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Apoptose/imunologia , Caspase 8/metabolismo , Proteína de Domínio de Morte Associada a Fas/metabolismo , Fosfoproteínas/metabolismo , Linfócitos T/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/imunologia , Animais , Western Blotting , Caspase 8/imunologia , Proteína de Domínio de Morte Associada a Fas/imunologia , Citometria de Fluxo , Humanos , Imunoprecipitação , Marcação In Situ das Extremidades Cortadas , Células Jurkat , Camundongos , Camundongos Knockout , Fosfoproteínas/imunologia , RNA Interferente Pequeno , Linfócitos T/imunologia , Transfecção , Receptor fas/imunologia , Receptor fas/metabolismo
10.
Trends Immunol ; 31(7): 260-9, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20576468

RESUMO

FAS-associated protein with death domain (FADD) is the key adaptor protein transmitting apoptotic signals mediated by the main death receptors (DRs). Besides being an essential instrument in cell death, FADD is also implicated in proliferation, cell cycle progression, tumor development, inflammation, innate immunity, and autophagy. Recently, many of these new functions of FADD were shown to be independent of DRs. Moreover, FADD function is dictated by protein localization and phosphorylation state. Thus, FADD is a crucial and unique controller of many essential cellular processes. The full understanding of the networks dictating the ultimate function of FADD may provide a new paradigm for other multifaceted proteins.


Assuntos
Proteína de Domínio de Morte Associada a Fas/imunologia , Animais , Apoptose , Humanos , Imunidade Inata , NF-kappa B/metabolismo , Transdução de Sinais , Linfócitos T/citologia , Linfócitos T/imunologia
11.
Curr Opin Pediatr ; 25(6): 722-9, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24240292

RESUMO

PURPOSE OF REVIEW: Several autoimmune lymphoproliferative syndromes have been described lately. We review here the main clinical and laboratory findings of these new disorders. RECENT FINDINGS: The prototypical autoimmune lymphoproliferative syndrome (ALPS) has had its diagnostic criteria modified, somatic mutations in RAS genes were found to cause an ALPS-like syndrome in humans, and mutations in a gene encoding a protein kinase C (PRKCD) were discovered to cause a syndrome of lymphoproliferation, autoimmunity and natural killer cell defect. SUMMARY: The recent discoveries shed light on the molecular pathways governing lymphocyte death, proliferation and immune tolerance in humans.


Assuntos
Apoptose/imunologia , Síndrome Linfoproliferativa Autoimune/diagnóstico , Genes ras , Tolerância Imunológica/imunologia , Inflamação/imunologia , Apoptose/genética , Síndrome Linfoproliferativa Autoimune/genética , Síndrome Linfoproliferativa Autoimune/imunologia , Autoimunidade/genética , Proliferação de Células , Proteína de Domínio de Morte Associada a Fas/imunologia , Feminino , Genes ras/genética , Genes ras/imunologia , Humanos , Tolerância Imunológica/genética , Inflamação/genética , Inflamação/patologia , Masculino , Mutação/imunologia , Proteína Quinase C-delta/imunologia , Transdução de Sinais/genética , Transdução de Sinais/imunologia
12.
Rheumatology (Oxford) ; 51(6): 964-75, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22253026

RESUMO

Inflammation is the principal hallmark of RA. Different pathways are implicated in the production of pro-inflammatory cytokines, the bona fide mediators of this inflammation. Among them are the TNF pathway and the IL-1 receptor/Toll-like receptor (IL-1R/TLR4) pathway. One of the potential negative regulators of IL-1R/TLR4 signalling is the Fas-associated death domain protein (FADD), which is the pivotal adaptor of the apoptotic signal mediated by death receptors of the TNF family. FADD can sequester myeloid differentiation primary response gene 88 (MyD88), the common adaptor of most TLRs, and hence hinder the activation of nuclear factor κB (NF-κB), the downstream transcription factor. We recently described a new regulatory mechanism of FADD expression, via the shedding of microvesicles, mediated by adenosine receptors. Interestingly, adenosine is found in high concentrations in the joints of RA patients and has been largely reported as a regulator of inflammation. This review discusses the possible link that could exist between the adenosine-dependent regulation of FADD in the inflammatory context of RA and the potential role of FADD as a therapeutic target in the treatment of RA. We will see that the modulation of FADD expression may be a double-edged sword by increasing apoptosis and at the same time limiting NF-κB activation.


Assuntos
Adenosina/metabolismo , Artrite Reumatoide/imunologia , Artrite Reumatoide/metabolismo , Proteína de Domínio de Morte Associada a Fas/imunologia , Proteína de Domínio de Morte Associada a Fas/metabolismo , Transdução de Sinais/imunologia , Humanos
13.
Blood ; 115(2): 265-73, 2010 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-19965651

RESUMO

Activation of the T cell-mediated immune response has been associated with changes in the expression of specific microRNAs (miRNAs). However, the role of miRNAs in the development of an effective immune response is just beginning to be explored. This study focuses on the functional role of miR-146a in T lymphocyte-mediated immune response and provides interesting clues on the transcriptional regulation of miR-146a during T-cell activation. We show that miR-146a is low in human naive T cells and is abundantly expressed in human memory T cells; consistently, miR-146a is induced in human primary T lymphocytes upon T-cell receptor (TCR) stimulation. Moreover, we identified NF-kB and c-ETS binding sites as required for the induction of miR-146a transcription upon TCR engagement. Our results demonstrate that several signaling pathways, other than inflammation, are influenced by miR-146a. In particular, we provide experimental evidence that miR-146a modulates activation-induced cell death (AICD), acting as an antiapoptotic factor, and that Fas-associated death domain (FADD) is a target of miR-146a. Furthermore, miR-146a enforced expression impairs both activator protein 1 (AP-1) activity and interleukin-2 (IL-2) production induced by TCR engagement, thus suggesting a role of this miRNA in the modulation of adaptive immunity.


Assuntos
Imunidade Adaptativa/fisiologia , Regulação da Expressão Gênica/fisiologia , Interleucina-2/biossíntese , Ativação Linfocitária/fisiologia , MicroRNAs/metabolismo , Linfócitos T/metabolismo , Morte Celular/fisiologia , Proteína de Domínio de Morte Associada a Fas/imunologia , Proteína de Domínio de Morte Associada a Fas/metabolismo , Humanos , Interleucina-2/imunologia , Células Jurkat , MicroRNAs/imunologia , Proteínas Proto-Oncogênicas c-ets/imunologia , Proteínas Proto-Oncogênicas c-ets/metabolismo , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Antígenos de Linfócitos T/metabolismo , Elementos de Resposta/fisiologia , Transdução de Sinais/fisiologia , Linfócitos T/citologia , Linfócitos T/imunologia , Fator de Transcrição AP-1/imunologia , Fator de Transcrição AP-1/metabolismo , Transcrição Gênica/fisiologia
14.
J Immunol ; 185(12): 7646-53, 2010 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-21068410

RESUMO

Apoptosis of oligodendrocytes (ODCs), the myelin-producing glial cells in the CNS, plays a central role in demyelinating diseases such as multiple sclerosis and experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis. To investigate the mechanism behind ODC apoptosis in EAE, we made use of conditional knockout mice lacking the adaptor protein FADD specifically in ODCs (FADD(ODC-KO)). FADD mediates apoptosis by coupling death receptors with downstream caspase activation. In line with this, ODCs from FADD(ODC-KO) mice were completely resistant to death receptor-induced apoptosis in vitro. In the EAE model, FADD(ODC-KO) mice followed an ameliorated clinical disease course in comparison with control littermates. Lymphocyte and macrophage infiltration into the spinal cord parenchyma was significantly reduced, as was the extent of demyelination and proinflammatory gene expression. Collectively, our data show that FADD is critical for ODC apoptosis and the development of autoimmune demyelinating disease.


Assuntos
Apoptose/imunologia , Encefalomielite Autoimune Experimental/imunologia , Proteína de Domínio de Morte Associada a Fas/imunologia , Deleção de Genes , Esclerose Múltipla/imunologia , Oligodendroglia/imunologia , Animais , Apoptose/genética , Encefalomielite Autoimune Experimental/genética , Encefalomielite Autoimune Experimental/metabolismo , Encefalomielite Autoimune Experimental/patologia , Proteína de Domínio de Morte Associada a Fas/biossíntese , Proteína de Domínio de Morte Associada a Fas/genética , Regulação da Expressão Gênica/genética , Regulação da Expressão Gênica/imunologia , Inflamação/genética , Inflamação/imunologia , Inflamação/metabolismo , Inflamação/patologia , Linfócitos/imunologia , Linfócitos/metabolismo , Linfócitos/patologia , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/patologia , Camundongos , Camundongos Knockout , Esclerose Múltipla/genética , Esclerose Múltipla/metabolismo , Esclerose Múltipla/patologia , Bainha de Mielina/imunologia , Bainha de Mielina/metabolismo , Bainha de Mielina/patologia , Oligodendroglia/metabolismo , Oligodendroglia/patologia , Medula Espinal/imunologia , Medula Espinal/metabolismo , Medula Espinal/patologia
15.
Lupus ; 20(1): 71-9, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21078760

RESUMO

Dysregulated neutrophil apoptosis may result in the development of autoimmune disease by contributing to nuclear autoantigen exposure, leading to autoantibody generation and a breakdown in immune tolerance. It has previously been shown that neutrophil apoptosis is increased in juvenile-onset systemic lupus erythematosus (JSLE). This study aims to investigate the pathways involved in JSLE serum-induced apoptosis. Caspases 3, 7-9, IAP1/2, XIAP and FADD mRNA levels and TRAIL R2, BID/tBID, caspase 8 and 9 protein expression were measured in neutrophils from JSLE patients (n = 14) and controls (n = 10). The mRNA levels of caspases 7-9 were significantly higher in JSLE neutrophils than in controls, whereas the mRNA levels of IAP1, IAP2 and XIAP were decreased (p < 0.05). A decrease in neutrophil apoptosis induced by JSLE serum was observed in the presence of caspase 8 and 9 inhibitors (p < 0.05), and the activity of caspases 8 and 9 increased over time. tBID protein expression increased following incubation with JSLE serum. These data focus specifically on the expression and activity of the main caspases in the intrinsic and extrinsic apoptotic pathways. Increased expression of factors involved in the downstream signalling of the extrinsic apoptotic pathway indicates a prominent involvement of this pathway in JSLE serum-induced apoptosis.


Assuntos
Apoptose/imunologia , Lúpus Eritematoso Sistêmico/imunologia , Adolescente , Proteína Agonista de Morte Celular de Domínio Interatuante com BH3/genética , Proteína Agonista de Morte Celular de Domínio Interatuante com BH3/imunologia , Inibidores de Caspase , Caspases/genética , Caspases/imunologia , Criança , Proteína de Domínio de Morte Associada a Fas/genética , Proteína de Domínio de Morte Associada a Fas/imunologia , Feminino , Humanos , Proteínas Inibidoras de Apoptose/genética , Proteínas Inibidoras de Apoptose/imunologia , Masculino , Neutrófilos/citologia , Neutrófilos/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/genética , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/imunologia , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/genética , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/imunologia
17.
Sheng Wu Gong Cheng Xue Bao ; 36(6): 1198-1208, 2020 Jun 25.
Artigo em Chinês | MEDLINE | ID: mdl-32597069

RESUMO

In this research, we studied the formation of Drosophila melanogaster FADD (Fas-associated death domain-containing protein) amyloid fiber and its influence on signal transduction in IMD (Immune deficiency) signaling pathway to better understand the regulation mechanism of Drosophila innate immune signaling pathway, which will provide reference for the immune regulation in other species. First, we purified dFADD protein expressed in Escherichia coli and performed Sulfur flavin T binding and transmission electron microscopy to identify the dFADD amyloid fibers formed in vitro. Then we investigated the formation of dFADD polymers in S2 cells using SDD-AGE and confocal microscope. We also constructed dFADD mutants to find out which domain is essential to fiber formation and its effect on IMD signal transduction. Our results revealed that dFADD could be polymerized to form amyloid fiber polymers in vitro and inside the cells. Formation of fibers relies on DED (Death-effector domain) domain of dFADD, since DED domain-deleted mutant existed as a monomer. Dual luciferase reporter assay showed that intact DED domain was required for the induction of downstream antimicrobial peptides, indicating that fiber formation was the key to IMD signal transduction. Our study revealed the role of dFADD in mediating the cascade between IMD and Dredd in the IMD signaling pathway by forming amyloid fibers, suggesting an evolutionarily conserved regulatory mechanism of innate immune signaling pathway.


Assuntos
Proteínas de Drosophila , Drosophila melanogaster , Imunidade Inata , Transdução de Sinais , Animais , Proteínas de Drosophila/biossíntese , Proteínas de Drosophila/imunologia , Drosophila melanogaster/imunologia , Proteína de Domínio de Morte Associada a Fas/biossíntese , Proteína de Domínio de Morte Associada a Fas/imunologia , Imunidade Inata/imunologia
18.
J Clin Invest ; 129(4): 1551-1565, 2019 02 25.
Artigo em Inglês | MEDLINE | ID: mdl-30694219

RESUMO

Across clinical trials, T cell expansion and persistence following adoptive cell transfer (ACT) have correlated with superior patient outcomes. Herein, we undertook a pan-cancer analysis to identify actionable ligand-receptor pairs capable of compromising T cell durability following ACT. We discovered that FASLG, the gene encoding the apoptosis-inducing ligand FasL, is overexpressed within the majority of human tumor microenvironments (TMEs). Further, we uncovered that Fas, the receptor for FasL, is highly expressed on patient-derived T cells used for clinical ACT. We hypothesized that a cognate Fas-FasL interaction within the TME might limit both T cell persistence and antitumor efficacy. We discovered that genetic engineering of Fas variants impaired in the ability to bind FADD functioned as dominant negative receptors (DNRs), preventing FasL-induced apoptosis in Fas-competent T cells. T cells coengineered with a Fas DNR and either a T cell receptor or chimeric antigen receptor exhibited enhanced persistence following ACT, resulting in superior antitumor efficacy against established solid and hematologic cancers. Despite increased longevity, Fas DNR-engineered T cells did not undergo aberrant expansion or mediate autoimmunity. Thus, T cell-intrinsic disruption of Fas signaling through genetic engineering represents a potentially universal strategy to enhance ACT efficacy across a broad range of human malignancies.


Assuntos
Transferência Adotiva , Engenharia Genética , Neoplasias Experimentais/terapia , Receptores de Antígenos Quiméricos , Transdução de Sinais/imunologia , Microambiente Tumoral/imunologia , Animais , Proteína Ligante Fas/genética , Proteína Ligante Fas/imunologia , Proteína de Domínio de Morte Associada a Fas/genética , Proteína de Domínio de Morte Associada a Fas/imunologia , Feminino , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Neoplasias Experimentais/genética , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/patologia , Receptores de Antígenos Quiméricos/genética , Receptores de Antígenos Quiméricos/imunologia , Receptores de Antígenos Quiméricos/uso terapêutico , Transdução de Sinais/genética , Microambiente Tumoral/genética , Receptor fas/genética , Receptor fas/imunologia
19.
FEBS J ; 286(3): 523-535, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30536547

RESUMO

Fas (CD95) signalling is best known for its role in apoptosis, however, recent reports have shown it to be involved in other cellular responses as well, including inflammation. Fas and its adaptor protein FADD are known to negatively regulate LPS-induced proinflammatory responses, but their role in LPS-induced type I interferon production is unknown. Here, we demonstrate that Fas engagement on macrophages, using an agonistic Fas antibody CH11, augments LPS-induced NF-κB responses, causing increased production of TNFα, IL-8, IL-6 and IL-12. Conversely, costimulation with both LPS and CH11 causes a significant reduction in the level of interferon-beta (IFNß) production. This differential effect involves the Fas adaptor FADD because while LPS-induced IL-6 production increased in FADD-/- murine embryonic fibroblasts, LPS-induced IFNß production was significantly reduced in these cells. Overexpression of a dominant negative form of FADD (FADD-DD) inhibits LPS-induced IFNß luciferase but not LPS-induced NF-κB luciferase. In contrast, overexpression of full-length FADD inhibited LPS-induced NF-κB luciferase activation but was seen to augment LPS-induced IFNß luciferase. Moreover, FADD-DD inhibits TRIF-, TRAM-, IKKε-, TBK-1- and TRAF3-induced IFNß luciferase production, with coimmunoprecipitation experiments demonstrating an interaction between FADD and TRIF. These data identify FADD as a novel component of the noncanonical Toll-like receptor 4/IFNß signalling pathway and demonstrate that both Fas and its adaptor FADD can differentially regulate the production of LPS-induced proinflammatory cytokines and type I interferons.


Assuntos
Proteína de Domínio de Morte Associada a Fas/genética , Interferon beta/genética , Lipopolissacarídeos/farmacologia , Macrófagos/efeitos dos fármacos , Receptor 4 Toll-Like/genética , Receptor fas/genética , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/imunologia , Proteínas Adaptadoras de Transporte Vesicular/genética , Proteínas Adaptadoras de Transporte Vesicular/imunologia , Animais , Anticorpos/farmacologia , Proteína de Domínio de Morte Associada a Fas/imunologia , Regulação da Expressão Gênica , Células HEK293 , Humanos , Quinase I-kappa B/genética , Quinase I-kappa B/imunologia , Interferon beta/imunologia , Interleucina-12/genética , Interleucina-12/imunologia , Interleucina-6/genética , Interleucina-6/imunologia , Interleucina-8/genética , Interleucina-8/imunologia , Células Jurkat , Macrófagos/citologia , Macrófagos/imunologia , Camundongos , NF-kappa B/genética , NF-kappa B/imunologia , Cultura Primária de Células , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/imunologia , Células RAW 264.7 , Transdução de Sinais , Células THP-1 , Fator 3 Associado a Receptor de TNF/genética , Fator 3 Associado a Receptor de TNF/imunologia , Receptor 4 Toll-Like/imunologia , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia , Receptor fas/antagonistas & inibidores , Receptor fas/imunologia
20.
Biotechnol Appl Biochem ; 50(Pt 3): 143-6, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18167119

RESUMO

FADD (Fas-associated death domain) has been widely expressed in various tissues and its expression has been recently demonstrated to correlate with tumour progression and prognosis. Currently, measurement of FADD expression mainly depends on Western-blot or immunohistochemical approaches. To develop a conventional sandwich ELISA avenue for the detection of FADD protein to supplement Western blotting or immunohistochemistry, a series of mAbs (monoclonal antibodies) specific for FADD protein, designated 3A3, 3F9, 3G4, 4B9, 4G1, 7A8, 7B8 and 7F4, were produced by fusing mouse s/p20 myeloma cells with the spleen cells of a mouse immunized with the Escherichia coli-expressed recombinant His(6)-FADD protein. On the basis of the characterization of these mAbs, purified 3F9 was selected as the capture antibody and the biotin-conjugated 3A3 was selected as the detection antibody in sandwich ELISA. The limit of detection for the ELISA was 0.3 ng of purified His(6)-FADD (FADD tagged with hexahistidine), and it could detect both recombinant and native human FADD protein. Furthermore, the positive reaction of the ELISA could be blocked by rabbit anti-FADD sera. All of these results indicated that the ELISA developed in the present paper could be a promising tool for detection of FADD protein.


Assuntos
Anticorpos Monoclonais/imunologia , Ensaio de Imunoadsorção Enzimática/métodos , Proteína de Domínio de Morte Associada a Fas/análise , Proteína de Domínio de Morte Associada a Fas/imunologia , Perfilação da Expressão Gênica/métodos , Rim/imunologia , Animais , Linhagem Celular , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Sensibilidade e Especificidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA